Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Am Soc Nephrol ; 31(7): 1522-1537, 2020 07.
Article in English | MEDLINE | ID: mdl-32503896

ABSTRACT

BACKGROUND: Mutations in the gene that encodes the lysosomal cystine transporter cystinosin cause the lysosomal storage disease cystinosis. Defective cystine transport leads to intralysosomal accumulation and crystallization of cystine. The most severe phenotype, nephropathic cystinosis, manifests during the first months of life, as renal Fanconi syndrome. The cystine-depleting agent cysteamine significantly delays symptoms, but it cannot prevent progression to ESKD and does not treat Fanconi syndrome. This suggests the involvement of pathways in nephropathic cystinosis that are unrelated to lysosomal cystine accumulation. Recent data indicate that one such potential pathway, lysosome-mediated degradation of autophagy cargoes, is compromised in cystinosis. METHODS: To identify drugs that reduce levels of the autophagy-related protein p62/SQSTM1 in cystinotic proximal tubular epithelial cells, we performed a high-throughput screening on the basis of an in-cell ELISA assay. We then tested a promising candidate in cells derived from patients with, and mouse models of, cystinosis, and in preclinical studies in cystinotic zebrafish. RESULTS: Of 46 compounds identified as reducing p62/SQSTM1 levels in cystinotic cells, we selected luteolin on the basis of its efficacy, safety profile, and similarity to genistein, which we previously showed to ameliorate other lysosomal abnormalities of cystinotic cells. Our data show that luteolin improves the autophagy-lysosome degradative pathway, is a powerful antioxidant, and has antiapoptotic properties. Moreover, luteolin stimulates endocytosis and improves the expression of the endocytic receptor megalin. CONCLUSIONS: Our data show that luteolin improves defective pathways of cystinosis and has a good safety profile, and thus has potential as a treatment for nephropathic cystinosis and other renal lysosomal storage diseases.


Subject(s)
Antioxidants/pharmacology , Cystinosis/drug therapy , Drug Evaluation, Preclinical/methods , Luteolin/pharmacology , RNA, Messenger/metabolism , Amino Acid Transport Systems, Neutral/genetics , Animals , Antioxidants/adverse effects , Apoptosis/drug effects , Autophagy/drug effects , Cells, Cultured , Cystinosis/metabolism , Disease Models, Animal , Endocytosis/drug effects , Humans , Kidney Tubules, Proximal/pathology , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Luteolin/adverse effects , Lysosomes/drug effects , Mice , Oxidative Stress/drug effects , Phenotype , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Zebrafish
2.
Nephrol Dial Transplant ; 35(7): 1163-1170, 2020 07 01.
Article in English | MEDLINE | ID: mdl-31006015

ABSTRACT

BACKGROUND: Zinc deficiency is commonly encountered in chronic kidney disease (CKD). The aims of this study were to assess whether zinc deficiency was related to increased renal excretion of zinc and to the progression of CKD. METHODS: Plasma and 24-h urinary zinc levels, urinary electrolytes and uromodulin were measured in 108 CKD patients and 81 individuals without CKD. Serum creatinine values were collected for 3 years to calculate the yearly change in estimated glomerular filtration rate (eGFR). Multivariable regression analysis was performed to assess the association between baseline zinc levels and yearly change in eGFR. RESULTS: CKD patients had lower circulating zinc levels and higher 24-h urinary zinc excretion than non-CKD participants (612.4 ± 425.9 versus 479.2 ± 293.0 µg/day; P = 0.02). Fractional excretion (FE) of zinc was higher and it significantly increased at more advanced CKD stages. Zinc FE was correlated negatively with 24-h urinary uromodulin excretion (r=-0.29; P < 0.01). Lower baseline plasma zinc levels were associated with a faster yearly decline of renal function in age, gender, diabetes and hypertension adjusted models, but this relationship was no longer significant when baseline eGFR or proteinuria were included. CONCLUSIONS: Zinc levels are lower in CKD, and not compensated by reduced renal zinc excretion. The inverse association between urinary zinc excretion and uromodulin possibly points to an impaired tubular activity, which could partly account for zinc imbalance in CKD. These data suggest that zinc status is associated with renal function decline, but further studies elucidating the underlying mechanisms and the potential role of zinc supplements in CKD are needed.


Subject(s)
Renal Insufficiency, Chronic/physiopathology , Zinc/blood , Zinc/deficiency , Case-Control Studies , Creatinine/blood , Disease Progression , Female , Follow-Up Studies , Glomerular Filtration Rate , Humans , Kidney Function Tests , Male , Middle Aged , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/urine , Uromodulin/urine
3.
Kidney Int ; 91(1): 24-33, 2017 01.
Article in English | MEDLINE | ID: mdl-28003083

ABSTRACT

Gitelman syndrome (GS) is a rare, salt-losing tubulopathy characterized by hypokalemic metabolic alkalosis with hypomagnesemia and hypocalciuria. The disease is recessively inherited, caused by inactivating mutations in the SLC12A3 gene that encodes the thiazide-sensitive sodium-chloride cotransporter (NCC). GS is usually detected during adolescence or adulthood, either fortuitously or in association with mild or nonspecific symptoms or both. The disease is characterized by high phenotypic variability and a significant reduction in the quality of life, and it may be associated with severe manifestations. GS is usually managed by a liberal salt intake together with oral magnesium and potassium supplements. A general problem in rare diseases is the lack of high quality evidence to inform diagnosis, prognosis, and management. We report here on the current state of knowledge related to the diagnostic evaluation, follow-up, management, and treatment of GS; identify knowledge gaps; and propose a research agenda to substantiate a number of issues related to GS. This expert consensus statement aims to establish an initial framework to enable clinical auditing and thus improve quality control of care.


Subject(s)
Bartter Syndrome/diagnosis , Chondrocalcinosis/etiology , Dietary Supplements , Gitelman Syndrome/diagnosis , Gitelman Syndrome/drug therapy , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Bartter Syndrome/blood , Bartter Syndrome/genetics , Bartter Syndrome/urine , Calcium/urine , Chloride Channels/genetics , Chondrocalcinosis/prevention & control , Consensus Development Conferences as Topic , Diagnosis, Differential , Genetic Testing , Gitelman Syndrome/complications , Gitelman Syndrome/genetics , Humans , Hypokalemia/blood , Hypokalemia/genetics , Magnesium/administration & dosage , Magnesium/blood , Magnesium/therapeutic use , Mutation , Phenotype , Potassium/administration & dosage , Potassium/blood , Potassium/therapeutic use , Practice Guidelines as Topic , Quality of Life , Rare Diseases/genetics , Sodium Chloride, Dietary/therapeutic use , Solute Carrier Family 12, Member 3/genetics , Ultrasonography
4.
Nephrol Dial Transplant ; 31(3): 337-48, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26908832

ABSTRACT

Recently, the European Medicines Agency approved the use of the vasopressin V2 receptor antagonist tolvaptan to slow the progression of cyst development and renal insufficiency of autosomal dominant polycystic kidney disease (ADPKD) in adult patients with chronic kidney disease stages 1-3 at initiation of treatment with evidence of rapidly progressing disease. In this paper, on behalf of the ERA-EDTA Working Groups of Inherited Kidney Disorders and European Renal Best Practice, we aim to provide guidance for making the decision as to which ADPKD patients to treat with tolvaptan. The present position statement includes a series of recommendations resulting in a hierarchical decision algorithm that encompasses a sequence of risk-factor assessments in a descending order of reliability. By examining the best-validated markers first, we aim to identify ADPKD patients who have documented rapid disease progression or are likely to have rapid disease progression. We believe that this procedure offers the best opportunity to select patients who are most likely to benefit from tolvaptan, thus improving the benefit-to-risk ratio and cost-effectiveness of this treatment. It is important to emphasize that the decision to initiate treatment requires the consideration of many factors besides eligibility, such as contraindications, potential adverse events, as well as patient motivation and lifestyle factors, and requires shared decision-making with the patient.


Subject(s)
Benzazepines/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/genetics , Societies, Medical , Adult , Antidiuretic Hormone Receptor Antagonists/therapeutic use , Disease Progression , Europe , Humans , Hyponatremia , Tolvaptan
5.
Kidney Int ; 82(10): 1121-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22718190

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is associated with a urine-concentrating defect attributed to renal cystic changes. As PKD genes are expressed in the brain, altered central release of arginine vasopressin could also play a role. In order to help determine this we measured central and nephrogenic components of osmoregulation in 10 adults and 10 children with ADPKD, all with normal renal function, and compared them to 20 age- and gender-matched controls. Overnight water deprivation caused a lower rise in urine osmolality in the patients with ADPKD than controls, reflecting an impaired release of vasopressin and a peripheral defect in the patients. The reactivity of plasma vasopressin to water deprivation, as found in controls, was blunted in the patients with the latter showing lower urine osmolality for the same range of plasma vasopressin. The maximal urine osmolality correlated negatively with total kidney volume. Defective osmoregulation was confirmed in the children with ADPKD but was unrelated to number of renal cysts or kidney size. Thus, patients with ADPKD have an early defect in osmoregulation, with a blunted release of arginine vasopressin. This reflects expression of polycystins in hypothalamic nuclei that synthesize vasopressin, and this should be considered when evaluating treatments targeting the vasopressin pathway in ADPKD.


Subject(s)
Hypothalamus/physiopathology , Kidney/physiopathology , Osmoregulation , Polycystic Kidney, Autosomal Dominant/physiopathology , Adolescent , Adult , Aged , Case-Control Studies , Chi-Square Distribution , Child , Child, Preschool , Female , Humans , Hypothalamus/metabolism , Kidney/metabolism , Kidney/pathology , Male , Middle Aged , Neurophysins/blood , Osmolar Concentration , Polycystic Kidney, Autosomal Dominant/blood , Polycystic Kidney, Autosomal Dominant/pathology , Polycystic Kidney, Autosomal Dominant/urine , Protein Precursors/blood , TRPP Cation Channels/metabolism , Time Factors , Vasopressins/blood , Water Deprivation , Young Adult
6.
Am J Physiol Renal Physiol ; 298(2): F365-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19940036

ABSTRACT

Renal tubular reabsorption is important for extracellular fluid homeostasis and much of this occurs via the receptor-mediated endocytic pathway. This pathway is disrupted in Dent's disease, an X-linked renal tubular disorder that is characterized by low-molecular-weight proteinuria, hypercalciuria, nephrolithiasis, and renal failure. Dent's disease is due to mutations of CLC-5, a chloride/proton antiporter, expressed in endosomes and apical membranes of renal tubules. Loss of CLC-5 function alters receptor-mediated endocytosis and trafficking of megalin and cubilin, although the underlying mechanisms remain to be elucidated. Here, we report that CLC-5 interacts with kinesin family member 3B (KIF3B), a heterotrimeric motor protein that facilitates fast anterograde translocation of membranous organelles. Using yeast two-hybrid, glutathione-S-transferase pull-down and coimmunoprecipitation assays, the COOH terminus of CLC-5 and the coiled-coil and globular domains of KIF3B were shown to interact. This was confirmed in vivo by endogenous coimmunoprecipitation of CLC-5 and KIF3B and codistribution with endosomal markers in mouse kidney fractions. Confocal live cell imaging in kidney cells further demonstrated association of CLC-5 and KIF3B, and transport of CLC-5-containing vesicles along KIF3B microtubules. KIF3B overexpression and underexpression, using siRNA, had reciprocal effects on whole cell chloride current amplitudes, CLC-5 cell surface expression, and endocytosis of albumin and transferrin. Clcn5(Y/-) mouse kidneys and isolated proximal tubular polarized cells showed increased KIF3B expression, whose effects on albumin endocytosis were dependent on CLC-5 expression. Thus, the CLC-5 and KIF3B interaction is important for CLC-5 plasma membrane expression and for facilitating endocytosis and microtubular transport in the kidney.


Subject(s)
Chloride Channels/metabolism , Endocytosis/physiology , Kidney/metabolism , Kinesins/metabolism , Microtubules/metabolism , Adult , Albumins/metabolism , Animals , COS Cells , Cell Line , Chloride Channels/physiology , Chlorocebus aethiops , DNA, Complementary , Down-Regulation , Drug Interactions , Electric Conductivity , Gene Library , Genetic Diseases, X-Linked/physiopathology , Humans , Kidney/cytology , Kidney Diseases/physiopathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mice , Mice, Knockout , Protein Interaction Domains and Motifs , Protein Transport , Two-Hybrid System Techniques , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 104(37): 14849-54, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17804801

ABSTRACT

The distal convoluted tubule (DCT) plays an essential role in the reabsorption of NaCl by the kidney, a process that can be inhibited by thiazide diuretics. Parvalbumin (PV), a Ca(2+)-binding protein that plays a role in muscle fibers and neurons, is selectively expressed in the DCT, where its role remains unknown. We therefore investigated the renal phenotype of PV knockout mice (Pvalb(-/-)) vs. wild-type (Pvalb(+/+)) littermates. PV colocalized with the thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC) in the early DCT. The Pvalb(-/-) mice showed increased diuresis and kaliuresis at baseline with higher aldosterone levels and lower lithium clearance. Acute furosemide administration increased diuresis and natriuresis/kaliuresis, but, surprisingly, did not increase calciuria in Pvalb(-/-) mice. NaCl supplementation of Pvalb(-/-) mice increased calciuria at baseline and after furosemide. The Pvalb(-/-) mice showed no significant diuretic response to hydrochlorothiazide, but an accentuated hypocalciuria. A decreased expression of NCC was detected in the early DCT of Pvalb(-/-) kidneys in the absence of ultrastructural and apoptotic changes. The PV-deficient mice had a positive Ca(2+) balance and increased bone mineral density. Studies in mouse DCT cells showed that endogenous NCC expression is Ca(2+)-dependent and can be modulated by the levels of PV expression. These results suggest that PV regulates the expression of NCC by modulating intracellular Ca(2+) signaling in response to ATP in DCT cells. They also provide insights into the Ca(2+)-sparing action of thiazides and the pathophysiology of distal tubulopathies.


Subject(s)
Diuretics/pharmacology , Kidney/metabolism , Parvalbumins/metabolism , Sodium Chloride Symporter Inhibitors/pharmacology , Sodium/metabolism , Aldosterone/pharmacology , Animals , Bone Density , Calcium/analysis , Calcium/urine , Cell Line, Transformed , Cells, Cultured , Diuresis , Furosemide/pharmacology , Immunohistochemistry , Kidney/drug effects , Kidney Tubules, Distal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Parvalbumins/blood , Parvalbumins/urine , Potassium/urine , Sodium/urine , Sodium Chloride Symporter Inhibitors/adverse effects , Sodium Chloride Symporters/physiology , Thiazides/pharmacology
8.
J Am Soc Nephrol ; 18(4): 1271-83, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17329572

ABSTRACT

Gitelman syndrome (GS) is a recessive salt-losing tubulopathy that is caused by mutations in the SLC12A3 gene that encodes the sodium-chloride co-transporter (NCC). GS is characterized by significant inter- and intrafamilial phenotype variability, with early onset and/or severe clinical manifestations in some patients. No correlations between the disease variability and the position/nature of SLC12A3 mutations have been investigated thus far. In this study, extensive mutational analyses of SLC12A3 were performed in 27 patients with GS, including genomic DNA sequencing, multiplex ligation-dependent probe amplification, cDNA analysis, and quantification of allele-specific transcripts, in parallel with functional analyses in Xenopus laevis oocytes and detailed phenotyping. Twenty-six SLC12A3 mutations were identified in 25 patients with GS, including eight novel (detection rate 80%). Transcript analysis demonstrated that splicing mutations of SLC12A3 lead to frameshifted mRNA subject to degradation by nonsense-mediated decay. Heterologous expression documented a novel class of NCC mutants with defective intrinsic transport activity. A subgroup of patients presented with early onset, growth retardation, and/or detrimental manifestations, confirming the potential severity of GS. The mutations that were associated with a severe presentation were the combination at least for one allele of a missplicing resulting in a truncated transcript that was downregulated by nonsense-mediated decay or a nonfunctional, cell surface-absent mutant. The most recurrent mutation on the second allele was a newly described NCC mutant that affected the functional properties of the co-transporter. These data suggest that the nature/position of SLC12A3 mutation, combined with male gender, is a determinant factor in the severity of GS and provide new insights in the underlying pathogenic mechanisms of the disease.


Subject(s)
Gitelman Syndrome/etiology , Mutation , Receptors, Drug/genetics , Symporters/genetics , Transcription, Genetic , Animals , DNA, Complementary/chemistry , Female , Genotype , Gitelman Syndrome/genetics , Humans , Phenotype , RNA, Messenger/analysis , Receptors, Drug/physiology , Reverse Transcriptase Polymerase Chain Reaction , Solute Carrier Family 12, Member 3 , Symporters/physiology , Xenopus
9.
Am J Kidney Dis ; 48(5): e73-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17059986

ABSTRACT

We report a case of Gitelman syndrome (GS) in a dizygotic twin who presented at 12 years of age with growth delay, metabolic alkalosis, hypomagnesemia and hypokalemia with inappropriate kaliuresis, and idiopathic intracranial hypertension with bilateral papilledema (pseudotumor cerebri). The patient, her twin sister, and her mother also presented with cerebral cavernous malformations. Based on the early onset and normocalciuria, Bartter syndrome was diagnosed first. However, mutation analysis showed that the proband is a compound heterozygote for 2 mutations in SLC12A3: a substitution of serine by leucine at amino acid position 555 (p.Ser555Leu) and a novel guanine to cytosine transition at the 5' splice site of intron 22 (c.2633+1G>C), providing the molecular diagnosis of GS. These mutations were not detected in 200 normal chromosomes and cosegregated within the family. Analysis of complementary DNA showed that the heterozygous nucleotide change c.2633+1G>C caused the appearance of 2 RNA molecules, 1 normal transcript and 1 skipping the entire exon 22 (r.2521_2634del). Supplementation with potassium and magnesium improved clinical symptoms and resulted in catch-up growth, but vision remained impaired. Three similar associations of Bartter syndrome/GS with pseudotumor cerebri were found in the literature, suggesting that electrolyte abnormalities and secondary aldosteronism may have a role in idiopathic intracranial hypertension. This study provides further evidence for the phenotypical heterogeneity of GS and its association with severe manifestations in children. It also shows the independent segregation of familial cavernomatosis and GS.


Subject(s)
Diseases in Twins/genetics , Gitelman Syndrome/genetics , Protein Splicing/genetics , Pseudotumor Cerebri/genetics , Receptors, Drug/genetics , Symporters/genetics , Alkalosis/genetics , Bartter Syndrome/genetics , Child , DNA Mutational Analysis , Diuretics , Female , Growth Disorders/genetics , Humans , Hypokalemia/genetics , Magnesium Deficiency/genetics , Magnetic Resonance Imaging , Mutation , Pedigree , Pseudotumor Cerebri/complications , Pseudotumor Cerebri/diagnosis , Solute Carrier Family 12, Member 3 , Syndrome
10.
Perit Dial Int ; 25 Suppl 3: S11-4, 2005 Feb.
Article in English | MEDLINE | ID: mdl-16048247

ABSTRACT

Acute peritonitis is the most frequent complication of peritoneal dialysis. Previous studies have suggested a major role for nitric oxide (NO) in the permeability changes and loss of ultrafiltration induced by acute peritonitis. In this study, we further investigated the potential role of NO in a mouse model of peritonitis induced by Escherichia coli Lipopolysaccharide (LPS). A 2-hour peritoneal equilibration test was performed in control and LPS-treated mice using 7% glucose dialysate supplemented or not with the NO synthase inhibitor NG-nitro-L-arginine methyl ester (L-NAME). The levels of NO metabolites in the dialysate were maximal 18 hours after LPS injection. At that time, acute peritonitis induced by LPS was reflected by an increased recruitment of leukocytes, an increased intraperitoneal release of interleukin-6, a significant increase in the peritoneal permeability for small solutes, a loss of sodium sieving, and a loss of ultrafiltration in comparison with controls. Addition of L-NAME in LPS-treated mice significantly reversed permeability modifications and prevented the release of NO metabolites into the dialysate. These results confirm that increased NO mediates permeability modifications during acute peritonitis, and illustrate the potential of mouse models to investigate the molecular mechanisms regulating peritoneal permeability.


Subject(s)
Escherichia coli , Lipopolysaccharides/adverse effects , Nitric Oxide Synthase/antagonists & inhibitors , Peritoneum/metabolism , Peritonitis/prevention & control , Acute Disease , Animals , Enzyme Inhibitors/administration & dosage , Infusions, Parenteral , Male , Mice , Models, Animal , NG-Nitroarginine Methyl Ester/administration & dosage , Nitric Oxide Synthase/immunology , Peritoneum/drug effects , Peritonitis/etiology , Peritonitis/physiopathology , Permeability
SELECTION OF CITATIONS
SEARCH DETAIL